Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Food as medicine: targeting the uraemic phenotype in chronic kidney disease

Abstract

The observation that unhealthy diets (those that are low in whole grains, fruits and vegetables, and high in sugar, salt, saturated fat and ultra-processed foods) are a major risk factor for poor health outcomes has boosted interest in the concept of ‘food as medicine’. This concept is especially relevant to metabolic diseases, such as chronic kidney disease (CKD), in which dietary approaches are already used to ameliorate metabolic and nutritional complications. Increased awareness that toxic uraemic metabolites originate not only from intermediary metabolism but also from gut microbial metabolism, which is directly influenced by diet, has fuelled interest in the potential of ‘food as medicine’ approaches in CKD beyond the current strategies of protein, sodium and phosphate restriction. Bioactive nutrients can alter the composition and metabolism of the microbiota, act as modulators of transcription factors involved in inflammation and oxidative stress, mitigate mitochondrial dysfunction, act as senolytics and impact the epigenome by altering one-carbon metabolism. As gut dysbiosis, inflammation, oxidative stress, mitochondrial dysfunction, premature ageing and epigenetic changes are common features of CKD, these findings suggest that tailored, healthy diets that include bioactive nutrients as part of the foodome could potentially be used to prevent and treat CKD and its complications.

Key points

  • The foodome is the pool of all of the compounds that are present in a food sample and/or in a biological system that is interacting with the investigated food.

  • A food-as-medicine approach could be used as a novel strategy to utilize bioactive nutrients to target the uraemic phenotype in chronic kidney disease.

  • Epigenetic alterations, gut dysbiosis, mitochondrial dysfunction, inflammation, oxidative stress and premature ageing are common features of the uraemic phenotype that could potentially be targeted using a food-as-medicine approach.

  • Gut dysbiosis is associated with inflammation and increased cardiovascular risk; prebiotics, probiotics, synbiotics and food components, including polyphenols, sugars and proteins, could alter the diversity of the gut microbiota and the production of uraemic toxins.

  • Senotherapeutic dietary compounds could potentially mitigate the effects of premature ageing in chronic kidney disease and associated complications, such as disturbed mitochondrial metabolism.

  • Natural bioactive compounds, including those found in turmeric, broccoli sprouts, berries, propolis and other foods, are potential nutritional therapeutic agents that could modulate the expression of pro-inflammatory transcription factors and the inflammasome.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Foods that could potentially influence DNA methylation.
Fig. 2: The effect of food intake on the gut microbiota in CKD.
Fig. 3: Mechanisms by which food might modulate premature ageing in CKD.
Fig. 4: Mechanisms by which food and nutrients might affect mitochondrial function.
Fig. 5: Nutrients that activate NRF2.
Fig. 6: The effect of diet on endogenous NO.

Similar content being viewed by others

References

  1. Global Burden of Diseas. Diet collaborators. health effects of dietary risks in 195 countries, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet 393, 1958–1972 (2019).

    Google Scholar 

  2. Srour, B. et al. Ultraprocessed food consumption and risk of type 2 diabetes among participants of the nutrinet-santé prospective cohort. JAMA Intern. Med. 180, 283–291 (2019).

    PubMed Central  Google Scholar 

  3. World Health Organization. WHO report on the global tobacco epidemic. WHO https://www.who.int/tobacco/global_report/2017/en/ (2017).

  4. Stenvinkel, P., Meyer, C. J., Block, G. A., Chertow, G. M., Shiels, P. G. Understanding the role of the cytoprotective transcription factor nuclear factor erythroid 2-related factor 2-lessons from evolution, the animal kingdom and rare progeroid syndromes. Nephrol. Dial. Transplant. https://doi.org/10.1093/ndt/gfz120 (2019).

  5. O’Neill, B. & Raggi, P. The ketogenic diet pros and cons. Atherosclerosis 292, 119–126 (2019).

    PubMed  Google Scholar 

  6. Carriazo, S. et al. Dietary care for ADPKD patients: current status and future directions. Nutrients 11, E1576 (2019).

    PubMed  Google Scholar 

  7. Torres, J. A. et al. Ketosis ameliorates renal cyst growth in polycystic kidney disease. Cell Metab. 30, 1007–1023 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. de Cabo, R. & Matsson, M. P. Effects of intermittent fasting on health, aging and disease. N. Engl. J. Med. 381, 2541–2551 (2019).

    PubMed  Google Scholar 

  9. Kandouz, S., Shendi, A. M., Zheng, Y., Sandeman, S. R. & Davenport, A. Reduced protein bound uraemic toxins in vegetarian kidney failure patients treated by haemodiafiltration. Hemodial. Int. 20, 610–617 (2016).

    PubMed  Google Scholar 

  10. Black, A. P. et al. Does low-protein diet influence the uremic toxin serum levels from the gut microbiota in nondialysis chronic kidney disease patients? J. Ren. Nutr. 28, 208–214 (2018).

    CAS  PubMed  Google Scholar 

  11. Saglimbene, V. M. et al. Fruit and vegetable intake and mortality in adults undergoing maintenance hemodialysis. Clin. J. Am. Soc. Nephrol. 14, 250–260 (2019).

    PubMed  PubMed Central  Google Scholar 

  12. Sharaf, El Din, U. A., Salem, M. M. & Abdulazim, D. O. Stop chronic kidney disease progression: time is approaching. World J. Nephrol. 5, 258–273 (2016).

    Google Scholar 

  13. Adair, K. E. & Bowden, R. G. Ameliorating chronic kidney disease using a whole food plant-based diet. Nutrients 12, 1007 (2020).

    CAS  PubMed Central  Google Scholar 

  14. Carrero, J. J. et al. Plant-based diets to manage the risks and complications of chronic kidney disease. Nat. Rev. Nephrol. 16, 525–542 (2020).

    PubMed  Google Scholar 

  15. Kim, H. et al. Plant-based diets and incident CKD and kidney function. Clin. J. Am. Soc. Nephrol. 14, 682–691 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Forouhi, N. G. & Unwin, N. Global diet and health: old questions, fresh evidence, and new horizons. Lancet. 393, 1916–1918 (2019).

    PubMed  Google Scholar 

  17. Stockler-Pinto, M. B. et al. Brazil nut (Bertholletia excelsa, H.B.K.) improves oxidative stress and inflammation biomarkers in hemodialysis patients. Biol. Trace Elem. Res. 158, 105–112 (2014).

    CAS  PubMed  Google Scholar 

  18. Khakimov, B. & Engelsen, S. B. Resveratrol in the foodomics era: 1:25,000. Ann. N. Y. Acad. Sci. 1403, 48–58 (2017).

    CAS  PubMed  Google Scholar 

  19. Khakimov, B., Gurdeniz, G. & Engelsen, S. B. Trends in the application of chemometrics to foodomics studies. Acta Aliment. 44, 4–31 (2015).

    CAS  Google Scholar 

  20. Hu, E. A. et al. Dietary patterns and risk of incident chronic kidney disease: the atherosclerosis risk in communities study. Am. J. Clin. Nutr. 110, 713–721 (2019).

    PubMed  PubMed Central  Google Scholar 

  21. Kelly, J. T. et al. Healthy dietary patterns and risk of mortality and ESRD in CKD: a meta-analysis of cohort studies. Clin. J. Am. Soc. Nephrol. 12, 272–279 (2017).

    PubMed  Google Scholar 

  22. Khoueiry, G. et al. Dietary intake in hemodialysis patients does not reflect a heart healthy diet. J. Ren. Nutr. 21, 438–447 (2011).

    CAS  PubMed  Google Scholar 

  23. Sussman, E. J., Singh, B., Clegg, D., Palmer, B. F. & Kalantar-Zadeh, K. Let them eat healthy: can emerging potassium binders help overcome dietary potassium restrictions in chronic kidney disease? J. Ren. Nutr. https://doi.org/10.1053/j.jrn.2020.01.022 (2020).

  24. Lehallier, B. et al. Undulating changes in human plasma proteome profiles across the lifespan. Nat. Med. 25, 1843–1850 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Clark, M. A., Springmann, M., Hill, J. & Tilman, D. Multiple health and environmental impacts of foods. Proc. Natl Acad. Sci. USA 116, 23357–23362 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Stenvinkel, P. The one health concept — the health of humans is intimately linked with the health of animals and a sustainable environment. J. Int. Med. 287, 223–225 (2020).

    CAS  Google Scholar 

  27. Stenvinkel, P. et al. A planetary health perspective for kidney disease. Kidney Int. 98, 261–265 (2020).

    PubMed  PubMed Central  Google Scholar 

  28. Dinkova-Kostova, A. T. & Abramov, A. Y. The emerging role of Nrf2 in mitochondrial function. Free. Radic. Biol. Med. 88, 179–188 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Fulop, G. et al. Nrf2 deficiency in aged mice exacerbates cellular senescence promoting cerebrovascular inflammation. Geroscience. 40, 513–521 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Mischke, M. & Plösch, T. The gut microbiota and their metabolites: potential implications for the host epigenome. Adv. Exp. Med. Biol. 902, 33–44 (2016).

    PubMed  Google Scholar 

  31. Shiels, P. G., McGuinness, D., Eriksson, M., Kooman, J. P. & Stenvinkel, P. The role of epigenetics in renal ageing. Nat. Rev. Nephrol. 13, 471–482 (2017).

    CAS  PubMed  Google Scholar 

  32. Shiels, P. G., Buchanan, S., Selman, C. & Stenvinkel, P. Allostatic load and ageing: linking the microbiome and nutrition with age-related health. Biochem. Soc. Trans. 47, 1165–1172 (2019).

    CAS  PubMed  Google Scholar 

  33. Witasp, A. et al. Current epigenetic aspects the clinical kidney researcher should embrace. Clin. Sci. 131, 1649–1667 (2017).

    Google Scholar 

  34. Larkin, B. P., Glastras, S. J., Chen, H., Pollock, C. A. & Saad, S. DNA methylation and the potential role of demethylating agents in prevention of progressive chronic kidney disease. FASEB J. 32, 5215–5226 (2018).

    CAS  PubMed  Google Scholar 

  35. O’Toole, P. W. & Shiels, P. G. The role of the microbiota in sedentary lifestyle disorders and ageing: lessons from the animal kingdom. J. Intern. Med. 287, 271–282 (2020).

    PubMed  Google Scholar 

  36. Kooman, J. P. et al. Inflammation and premature aging in advanced chronic kidney disease. Am. J. Physiol. Ren. Physiol. 313, F938–F950 (2017).

    Google Scholar 

  37. Cañadas-Garre, M., Anderson, K., McGoldrick, J., Maxwell, A. P. & McKnight, A. J. Genomic approaches in the search for molecular biomarkers in chronic kidney disease. J. Transl. Med. 16, 292 (2018).

    PubMed  PubMed Central  Google Scholar 

  38. Wing, M. R. et al. Chronic renal insufficiency cohort (CRIC) study. DNA methylation profile associated with rapid decline in kidney function: findings from the CRIC study. Nephrol. Dial. Transplant. 29, 864–872 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Suliman, M. E., Bárány, P., Kalantar-Zadeh, K., Lindholm, B. & Stenvinkel, P. Homocysteine in uraemia — a puzzling and conflicting story. Nephrol. Dial. Transplant. 20, 16–21 (2005).

    PubMed  Google Scholar 

  40. Ingrosso, D. et al. Folate treatment and unbalanced methylation and changes of allelic expression induced by hyperhomocysteinaemia in patients with uraemia. Lancet. 361, 1693–1699 (2003).

    CAS  PubMed  Google Scholar 

  41. McGuinness, D. et al. A molecular signature for delayed graft function. Aging Cell 17, e12825 (2018).

    PubMed  PubMed Central  Google Scholar 

  42. Stenvinkel, P. et al. Impact of inflammation on epigenetic DNA methylation – a novel risk factor for cardiovascular disease? J. Intern. Med. 261, 488–499 (2007).

    CAS  PubMed  Google Scholar 

  43. McGuinness, D. et al. Identification of molecular markers of delayed graft function based on the regulation of biological ageing. PLoS ONE 11, e0146378 (2016).

    PubMed  PubMed Central  Google Scholar 

  44. Chu, A. Y. et al. Epigenome-wide association studies identify DNA methylation associated with kidney function. Nat. Commun. 8, 1286 (2017).

    PubMed  PubMed Central  Google Scholar 

  45. Crider, K. S., Yang, T. P., Berry, R. J. & Bailey, L. B. Folate and DNA methylation: a review of molecular mechanisms and the evidence for folate’s role. Adv. Nutr. 3, 21–38 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Friso, S., Udali, S., De Santis, D. & Choi, S. W. One-carbon metabolism and epigenetics. Mol. Asp. Med. 24, 28–36 (2017).

    Google Scholar 

  47. Chu, D. M., Wahlqvist, M. L., Chang, H. Y., Yeh, N. H. & Lee, M. S. Choline and betaine food sources and intakes in Taiwanese. Asia Pac. J. Clin. Nutr. 21, 547–557 (2012).

    CAS  PubMed  Google Scholar 

  48. Mafra, D. et al. Methyl donor nutrients in chronic kidney disease: impact on the epigenetic landscape. J. Nutr. 149, 372–380 (2019).

    PubMed  Google Scholar 

  49. Clifford, T., Howatson, G., West, D. J. & Stevenson, E. J. The potential benefits of red beetroot supplementation in health and disease. Nutrients 14, 2801–2822 (2015).

    Google Scholar 

  50. Du, J. et al. Betaine supplementation enhances lipid metabolism and improves insulin resistance in mice fed a high-fat diet. Nutrients 10, E131 (2018).

    PubMed  Google Scholar 

  51. Missailidis, C. et al. Serum trimethylamine-N-oxide is strongly related to renal function and predicts outcome in chronic kidney disease. PLoS ONE 11, e0141738 (2016).

    PubMed  PubMed Central  Google Scholar 

  52. Lozupone, C. A., Stombaugh, J. I., Gordon, J. I., Jansson, J. K. & Knight, R. Diversity, stability and resilience of the human gut microbiota. Nature 489, 220–230 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Tang, W. H., Kitai, T. & Hazen, S. L. Gut microbiota in cardiovascular health and disease. Circ. Res. 120, 1183–1196 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Meijers, B., Evenepoel, P. & Anders, H. J. Intestinal microbiome and fitness in kidney disease. Nat. Rev. Nephrol. 15, 531–545 (2019).

    PubMed  Google Scholar 

  55. Vaziri, N. D., Zhao, Y. Y. & Pahl, M. V. Altered intestinal microbial flora and impaired epithelial barrier structure and function in CKD: the nature, mechanisms, consequences and potential treatment. Nephrol. Dial. Transplant. 31, 737–746 (2015).

    PubMed  Google Scholar 

  56. Al-Khodor, S. & Shatat, I. F. Gut microbiome and kidney disease: a bidirectional relationship. Pediatr. Nephrol. 32, 921–931 (2017).

    PubMed  Google Scholar 

  57. De Filippo, C. et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc. Natl Acad. Sci. USA 107, 14691–14696 (2010).

    PubMed  PubMed Central  Google Scholar 

  58. Wu, G. D. et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 334, 105–108 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Hida, M. et al. Inhibition of the accumulation of uremic toxins in the blood and their precursors in the feces after oral administration of Lebenin, a lactic acid bacteria preparation, to uremic patients undergoing hemodialysis. Nephron 74, 349–355 (1996).

    CAS  PubMed  Google Scholar 

  60. Taki, K., Takayama, F. & Niwa, T. Beneficial effects of Bifidobacteria in a gastroresistant seamless capsule on hyperhomocysteinemi a in hemodialysis patients. J. Ren. Nutr. 15, 77–80 (2005).

    PubMed  Google Scholar 

  61. Ranganathan, N. et al. Pilot study of probiotic dietary supplementation for promoting healthy kidney function in patients with chronic kidney disease. Adv. Ther. 27, 634–647 (2010).

    PubMed  Google Scholar 

  62. Miranda Alatriste, P. V., Urbina Arronte, R., Gomez Espinosa, C. O. & Espinosa Cuevas, M. de L. Effect of probiotics on human blood urea levels in patients with chronic renal failure. Nutr. Hosp. 29, 582–590 (2014).

    PubMed  Google Scholar 

  63. Natarajan, R. et al. Randomized controlled trial of strain-specific probiotic formulation (Renadyl) in dialysis patients. Biomed. Res. Int. 2014, 568571 (2014).

    PubMed  PubMed Central  Google Scholar 

  64. Borges, N. A. et al. Probiotic supplementation in chronic kidney disease: a double-blind, randomized, placebo-controlled trial. J. Ren. Nutr. 28, 28–36 (2018).

    CAS  PubMed  Google Scholar 

  65. Singh, S. P., Jadaun, J. S., Narnoliya, L. K. & Pandey, A. Prebiotic oligosaccharides: special focus on fructooligosaccharides, its biosynthesis and bioactivity. Appl. Biochem. Biotechnol. 183, 613–635 (2017).

    CAS  PubMed  Google Scholar 

  66. Walker, A. W. et al. Dominant and diet-responsive groups of bacteria within the human colonic microbiota. ISME J. 5, 220–230 (2011).

    CAS  PubMed  Google Scholar 

  67. Lecerf, J. M. et al. Xylo-oligosaccharide (XOS) in combination with inulin modulates both the intestinal environment and imune status in healthy subjects, while XOS alone only shows prebiotic properties. Br. J. Nutr. 108, 1847–1858 (2012).

    CAS  PubMed  Google Scholar 

  68. Graf, D. et al. Contribution of diet to the composition of the human gut microbiota. Microb. Ecol. Health Dis. 26, 26164 (2015).

    PubMed  Google Scholar 

  69. Upadhyaya, B. et al. Impact of dietary resistant starch type 4 on human gut microbiota and immunometabolic functions. Sci. Rep. 6, 28797 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Poesen, R. et al. The Influence of CKD on Colonic Microbial Metabolism. J. Am. Soc. Nephrol. 27, 1389–1399 (2016).

    CAS  PubMed  Google Scholar 

  71. Khosroshahi, H. T. et al. Effect of high amylose resistant starch (HAMRS2) supplementation on biomarkers of inflammation and oxidative stress in hemodialysis patients: a randomized clinical trial. Hemodial. Int. 22, 492–500 (2018).

    Google Scholar 

  72. Esgalhado, M. et al. Resistant starch supplementation improve inflammatory and oxidative stress biomarkers and uremic toxins levels in hemodialysis patients? A pilot randomized controlled trial. Food Funct. 13, 6508–6516 (2018).

    Google Scholar 

  73. Khosroshahi, H. T. et al. The effect of lactulose supplementation on fecal microflora of patients with chronic kidney disease: a randomized clinical trial. J. Renal Inj. Prev. 5, 162–167 (2016).

    Google Scholar 

  74. Gibson, G. R. et al. The international scientific association for probiotics and prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 14, 491–502 (2017).

    PubMed  Google Scholar 

  75. Tsai, Y. L. et al. Probiotics, prebiotics and amelioration of diseases. J. Biomed. Sci. 26, 3 (2019).

    PubMed  PubMed Central  Google Scholar 

  76. Al-Sheraji, S. H. et al. Prebiotics as functional foods: a review. J. Func. Foods. 5, 1542–1553 (2013).

    CAS  Google Scholar 

  77. Moraes, C., Borges, N. A. & Mafra, D. Resistant starch for modulation of gut microbiota: Promising adjuvant therapy for chronic kidney disease patients? Eur. J. Nutr. 55, 1813–1821 (2016).

    CAS  PubMed  Google Scholar 

  78. Cruz-Mora, J. et al. Effects of a symbiotic on gut microbiota in Mexican patients with end-stage renal disease. J. Ren. Nutr. 24, 330–335 (2014).

    PubMed  Google Scholar 

  79. Dehghani, H. H. F., Mozaffari-Khosravi, H., Nouri-Majelan, N. & Dehghani, A. Synbiotic supplementations for azotemia in patients with chronic kidney disease: a randomized controlled trial. Iran. J. Kidney Dis. 10, 351–357 (2016).

    PubMed  Google Scholar 

  80. Rossi, M. et al. Synbiotics easing renal failure by improving gut microbiology (SYNERGY): a randomized trial. Clin. J. Am. Soc. Nephrol. 11, 223–231 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. McFarlane, C., Ramos, C. I., Johnson, D. W. & Campbell, K. L. Prebiotic, probiotic, and synbiotic supplementation in chronic kidney disease: a systematic review and meta-analysis. J. Ren. Nutr. 29, 209–220 (2019).

    CAS  PubMed  Google Scholar 

  82. Rinninella, E. et al. The role of diet, micronutrients and the gut microbiota in age-related macular degeneration: new perspectives from the gut–retina axis. Nutrients 10, 1677 (2018).

    PubMed Central  Google Scholar 

  83. Rosas-Villegas, A. et al. Differential effect of sucrose and fructose in combination with a high fat diet on intestinal microbiota and kidney oxidative stress. Nutrients 9, E393 (2017).

    PubMed  Google Scholar 

  84. Do, M. H., Lee, E., Oh, M. J., Kim, Y. & Park, H. Y. High-glucose or -fructose diet cause changes of the gut microbiota and metabolic disorders in mice without body weight change. Nutrients 10, 761 (2018).

    PubMed Central  Google Scholar 

  85. Rysz, J., Franczyk, B., Ciałkowska-Rysz, A. & Gluba-Brzózka, A. The effect of diet on the survival of patients with chronic kidney disease. Nutrients 9, E495 (2017).

    PubMed  Google Scholar 

  86. Scott, K. P., Gratz, S. W., Sheridan, P. O., Flint, H. J. & Duncan, S. H. The influence of diet on the gut microbiota. Pharmacol. Res. 69, 52–60 (2013).

    CAS  PubMed  Google Scholar 

  87. Ercolini, D. & Fogliano, V. Food design to feed the human gut microbiota. J. Agric. Food Chem. 66, 3754–3758 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Evenepoel, P., Meijers, B. K., Bammens, B. R. & Verbeke, K. Uremic toxins originating from colonic microbial metabolism. Kidney Int. Suppl. 114, S12–S19 (2009).

    CAS  Google Scholar 

  89. Mafra, D., Barros, A. F. & Fouque, D. Dietary protein metabolism by gut microbiota and its consequences for chronic kidney disease patients. Future Microbiol. 8, 1317–1323 (2013).

    CAS  PubMed  Google Scholar 

  90. Swain Ewald, H. A. & Ewald, P. W. Natural selection, the microbiome, and public health. Yale J. Biol. Med. 91, 445–455 (2018).

    PubMed  PubMed Central  Google Scholar 

  91. Nyangale, E. P., Mottram, D. S. & Gibson, G. R. J. Gut microbial activity, implications for health and disease: the potential role of metabolite analysis. Proteome Res. 11, 5573–5585 (2012).

    CAS  Google Scholar 

  92. Madsen, L., Myrmel, L. S., Fjære, E., Liaset, B. & Kristiansen, K. Links between dietary protein sources, the gut microbiota, and obesity. Front. Physiol. 8, 1047 (2017).

    PubMed  PubMed Central  Google Scholar 

  93. Zhao, J., Zhang, X., Liu, H., Brown, M. A. & Qiao, S. Dietary protein and gut microbiota composition and function. Curr. Protein Pept. Sci. 20, 145–154 (2019).

    CAS  PubMed  Google Scholar 

  94. Song, M. & Chan, A. T. Diet, gut microbiota, and colorectal cancer prevention: a review of potential mechanisms and promising targets for future research. Curr. Colorectal Cancer Rep. 13, 429–439 (2017).

    PubMed  PubMed Central  Google Scholar 

  95. Mafra, D. et al. Red meat intake in chronic kidney disease patients: two sides of the coin. Nutrition 46, 26–32 (2018).

    CAS  PubMed  Google Scholar 

  96. Zhu, Y. et al. Beef, chicken, and soy proteins in diets induce different gut microbiota and metabolites in rats. Front. Microbiol. 8, 1395 (2017).

    PubMed  PubMed Central  Google Scholar 

  97. Ge, Y. et al. Effect of industrial trans-fatty acids-enriched diet on gut microbiota of C57BL/6 mice. Eur. J. Nutr. 58, 2625–2638 (2018).

    PubMed  Google Scholar 

  98. Wisniewski, P. J., Dowden, R. A. & Campbell, S. C. Role of dietary lipids in modulating inflammation through the gut microbiota. Nutrients 11, 117 (2019).

    CAS  PubMed Central  Google Scholar 

  99. Wan, Y. et al. Effects of dietary fat on gut microbiota and faecal metabolites, and their relationship with cardiometabolic risk factors: a 6-month randomised controlled-feeding trial. Gut 68, 1417–1429 (2019).

    CAS  PubMed  Google Scholar 

  100. Lam, Y. Y. et al. Effects of dietary fat profile on gut permeability and microbiota and their relationships with metabolic changes in mice. Obesity 23, 1429–1439 (2015).

    CAS  PubMed  Google Scholar 

  101. Costantini, L., Molinari, R., Farinon, B. & Merendino, N. Impact of omega-3 fatty acids on the gut microbiota. Int. J. Mol. Sci. 18, 2645 (2017).

    PubMed Central  Google Scholar 

  102. Martín-Peláez, S. et al. Effect of virgin olive oil and thyme phenolic compounds on blood lipid profile: Implications of human gut microbiota. Eur. J. Nutr. 56, 119–131 (2017).

    PubMed  Google Scholar 

  103. Prieto, I. et al. Influence of a diet enriched with virgin olive oil or butter on mouse gut microbiota and its correlation to physiological and biochemical parameters related to metabolic syndrome. PLoS ONE 13, e0190368 (2018).

    PubMed  PubMed Central  Google Scholar 

  104. Martínez, N. et al. Refined versus extra virgin olive oil high-fat diet impact on intestinal microbiota of mice and its relation to different physiological variables. Microorganisms 7, 61 (2019).

    PubMed Central  Google Scholar 

  105. Sarmugam, R. & Worsley, A. Current levels of salt knowledge: a review of the literature. Nutrients 6, 5534–5559 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Hu, J. et al. Enteric dysbiosis-linked gut barrier disruption triggers early renal injury induced by chronic high salt feeding in mice. Exp. Mol. Med. 49, e370 (2017).

    PubMed  PubMed Central  Google Scholar 

  107. Vazquez-Gutierrez, P. et al. Bifidobacteria strains isolated from stools of iron deficient infants can efficiently sequester iron. BMC Microbiol. 15, 3 (2015).

    PubMed  PubMed Central  Google Scholar 

  108. Richard, S. A. et al. Zinc and iron supplementation and malaria, diarrhea, and respiratory infections in children in the peruvian amazon. Am. J. Trop. Med. Hyg. 75, 126–132 (2006).

    CAS  PubMed  Google Scholar 

  109. Chang, S. et al. Supplementing iron and zinc: double blind, randomized evaluation of separate or combined delivery. Eur. J. Clin. Nutr. 64, 153–160 (2010).

    CAS  PubMed  Google Scholar 

  110. Nchito, M., Friis, H., Michaelsen, K. F., Mubila, L. & Olsen, A. Iron supplementation increases small intestine permeability in primary schoolchildren in Lusaka, Zambia. Trans. R. Soc. Trop. Med. Hyg. 100, 791–794 (2006).

    CAS  PubMed  Google Scholar 

  111. Zimmermann, M. B. et al. The effects of iron fortification on the gut microbiota in African children: a randomized controlled trial in Côte d’Ivoire. Am. J. Clin. Nutr. 92, 1406–1415 (2010).

    CAS  PubMed  Google Scholar 

  112. Kortman, G. A. et al. Microbial metabolism shifts towards an adverse profile with supplementary iron in the TIM-2 in vitro model of the human colon. Front. Microbiol. 6, 1481 (2016).

    PubMed  PubMed Central  Google Scholar 

  113. Chiang, C. K., Tanaka, T., Inagi, R., Fujita, T. & Nangaku, M. Indoxyl sulfate, a representative uremic toxin, suppresses erythropoietin production in a HIF-dependent manner. Lab. Invest. 91, 1564–1571 (2011).

    CAS  PubMed  Google Scholar 

  114. Bonan, N. B., Steiner, Y. M. & Kuntsevich, V. Uremic toxicity-induced eryptosis and monocyte modulation: the erythrophagocytosis as a novel pathway to renal anemia. Blood Purif. 41, 317–323 (2016).

    CAS  PubMed  Google Scholar 

  115. Kortman, G. A. M., Reijnders, D. & Swinkels, D. W. Oral iron supplementation: Potential implications for the gut microbiome and metabolome in patients with CKD. Hemodial. Int. 21, S28–S36 (2017).

    PubMed  Google Scholar 

  116. Bondonno, N. P. et al. Flavonoid intake is associated with lower mortality in the Danish diet cancer and health cohort. Nat. Commun. 10, 3651 (2019).

    PubMed  PubMed Central  Google Scholar 

  117. Ozdal, T. et al. The reciprocal interactions between polyphenols and gut microbiota and effects on bioaccessibility. Nutrients 8, 78 (2016).

    PubMed  PubMed Central  Google Scholar 

  118. Singh, R. et al. Enhancement of the gut barrier integrity by a microbial metabolite through the Nrf2 pathway. Nat. Commun. 10, 89 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Tzounis, X. et al. Prebiotic evaluation of cocoa-derived flavanols in healthy humans by using a randomized, controlled, double-blind, crossover intervention study. Am. J. Clin. Nutr. 93, 62–72 (2011).

    CAS  PubMed  Google Scholar 

  120. Lee, S. et al. Blueberry supplementation influences the gut microbiota, inflammation, and insulin resistance in high-fat-diet-fed rats. J. Nutr. 148, 209–219 (2018).

    PubMed  PubMed Central  Google Scholar 

  121. Wu, W.-K. et al. Dietary allicin reduces transformation of L-carnitine to TMAO through impact on gut microbiota. J. Func. Foods 15, 408–417 (2015).

    CAS  Google Scholar 

  122. Chen, M. L. et al. Resveratrol attenuates trimethylamine-N-oxide (TMAO)-induced atherosclerosis by regulating TMAO synthesis and bile acid metabolism via remodeling of the gut microbiota. MBio 7, e02210–e02215 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Cardona, F., Andrés-Lacueva, C., Tulipani, S., Tinahones, F. J. & Queipo-Ortuño, M. I. Benefits of polyphenols on gut microbiota and implications in human health. J. Nutr. Biochem. 24, 1415–1422 (2013).

    CAS  PubMed  Google Scholar 

  124. Pfeffer, M., Ziesenitz, S. C. & Siebert, G. Acesulfame K cyclamate and saccharin inhibit the anaerobic fermentation of glucose by intestinal bacteria. Z. Ernahrungswiss. 24, 231–235 (1985).

    CAS  PubMed  Google Scholar 

  125. Wang, Q. P., Browman, D., Herzog, H. & Neely, G. G. Non-nutritive sweeteners possess a bacteriostatic effect and alter gut microbiota in mice. PLoS ONE 13, e0199080 (2018).

    PubMed  PubMed Central  Google Scholar 

  126. Suez, J. et al. Artificial sweeteners induce glucose intolerance by altering the gut microbiota. Nature 514, 181–186 (2014).

    CAS  PubMed  Google Scholar 

  127. Abou-Donia, M. B., El-Masry, E. M., Abdel-Rahman, A. A., McLendon, R. E. & Schiffman, S. S. Splenda alters gut microflora and increases intestinal p-glycoprotein and cytochrome p-450 in male rats. J. Toxicol. Environ. Health A 71, 1415–1429 (2008).

    CAS  PubMed  Google Scholar 

  128. Daly, K., Darby, A. C. & Shirazi-Beechey, S. P. Low calorie sweeteners and gut microbiota. Physiol. Behav. 164, 494–500 (2016).

    CAS  PubMed  Google Scholar 

  129. Pepino, M. Y. Metabolic effects of non-nutritive sweeteners. Physiol. Behav. 152, 450–455 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Martínez-Carrillo, B. E. et al. Effect of chronic consumption of sweeteners on microbiota and immunity in the small intestine of young mice. Int. J. Food Sci. 2019, 9619020 (2019).

    PubMed  PubMed Central  Google Scholar 

  131. Garcia-Mantrana, I., Selma-Royo, M., Alcantara, C. & Collado, M. C. Shifts on gut microbiota associated to Mediterranean diet adherence and specific dietary intakes on general adult population. Front. Microbiol. 9, 890 (2018).

    PubMed  PubMed Central  Google Scholar 

  132. Bischoff, S. Microbiota and aging. Curr. Opin. Clin. Nutr. Metab. Care. 19, 26–30 (2016).

    CAS  PubMed  Google Scholar 

  133. Van Deursen, J. M. The role of senescent cells in ageing. Nature 509, 439–446 (2014).

    PubMed  PubMed Central  Google Scholar 

  134. Ferrucci, L. & Fabbri, E. Inflammageing: chronic inflammation in ageing, cardiovascular disease, and frailty. Nat. Rev. Cardiol. 15, 505–522 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Storer, M. et al. Senescence is a developmental mechanism that contributes to embryonic growth and patterning. Cell 155, 1119–1130 (2013).

    CAS  PubMed  Google Scholar 

  136. Munoz-Espin, D. et al. Programmed cell senescence during mammalian embryonic development. Cell 155, 1104–1118 (2013).

    CAS  PubMed  Google Scholar 

  137. Demaria, M. et al. An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA. Dev. Cell 31, 722–733 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Mosteiro, L. et al. Tissue damage and senescence provide critical signals for cellular reprogramming in vivo. Science 354, aaf4445 (2016).

    PubMed  Google Scholar 

  139. Coppé, J. P. et al. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 6, 2853–2868 (2008).

    PubMed  Google Scholar 

  140. Biran, A. et al. Quantitative identification of senescent cells in aging and disease. Aging Cell. 16, 661–671 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Kirkland, J. L. & Tchkonia, T. Cellular senescence: a translational perspective. EBioMedicine 21, 21–28 (2017).

    PubMed  PubMed Central  Google Scholar 

  142. de Kok, M. J. et al. The neglectable impact of delayed graft function on long-term graft survival in kidneys donated after circulatory death associates with superior organ resilience. Ann. Surg. 270, 877–883 (2019).

    PubMed  Google Scholar 

  143. Kooman, J. P., Kotanko, P., Schols, A. M., Shiels, P. G. & Stenvinkel, P. Chronic kidney disease and premature ageing. Nat. Rev. Nephrol. 10, 732–742 (2014).

    CAS  PubMed  Google Scholar 

  144. Hayflick, L. & Moorhead, P. S. The serial cultivation of human diploid cell strains. Exp. Cell. Res. 25, 585–621 (1961).

    CAS  PubMed  Google Scholar 

  145. Teo, Y. V. et al. Notch signaling mediates secondary senescence. Cell Rep. 27, 997–1007.e5 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Robinson, M. W. et al. Non cell autonomous upregulation of CDKN2 transcription linked to progression of chronic hepatitis C disease. Aging Cell 12, 1141–1143 (2013).

    CAS  PubMed  Google Scholar 

  147. Sturmlechner, I., Durik, M., Sieben, C. J., Baker, D. J. & van Deursen, J. M. Cellular senescence in renal ageing and disease. Nat. Rev. Nephrol. 13, 77–89 (2017).

    CAS  PubMed  Google Scholar 

  148. Palmer, A. K., Gustafson, B., Kirkland, J. L. & Smith, U. Cellular senescence: at the nexus between ageing and diabetes. Diabetologia 62, 1835–1841 (2019).

    PubMed  PubMed Central  Google Scholar 

  149. Stenvinkel, P. et al. CDKN2A/p16INK4a expression is associated with vascular progeria in chronic kidney disease. Aging 9, 494–507 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Tchkonia, T. & Kirkland, J. L. Aging, cell senescence, and chronic disease: emerging therapeutic strategies. JAMA 320, 1319–1320 (2018).

    PubMed  Google Scholar 

  151. Gurău, F. et al. Anti-senescence compounds: a potential nutraceutical approach to healthy aging. Ageing Res. Rev. 46, 14–31 (2018).

    PubMed  Google Scholar 

  152. Li, W. Emerging senolytic agents derived from natural products. Mech. Ageing Dev. 181, 1–6 (2019).

    CAS  PubMed  Google Scholar 

  153. Senger, D. R., Li, D., Jaminet, S.-C. & Cao, S. Activation of the Nrf2 cell defense pathway by ancient foods: disease prevention by important molecules and microbes lost from the modern Western diet. PLoS ONE 11, e0148042 (2016).

    PubMed  PubMed Central  Google Scholar 

  154. Gómez-Linton, D. R. et al. Some naturally occurring compounds that increase longevity and stress resistance in model organisms of aging. Biogerontology 20, 583–603 (2019).

    PubMed  Google Scholar 

  155. He, J. et al. The resistant effect of SIRT1 in oxidative stress-induced senescence of rat nucleus pulposus cell is regulated by Akt-FoxO1 pathway. Biosci. Rep. 39, BSR20190112 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Man, A. W. C., Li, H. & Xia, N. The role of sirtuin1 in regulating endothelial function, arterial remodeling and vascular aging. Front. Physiol. 10, 1173 (2019).

    PubMed  PubMed Central  Google Scholar 

  157. Shiels, P. G. et al. Accelerated telomere attrition is associated with relative household income, diet and inflammation in the pSoBid cohort. PLoS ONE 6, e22521 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. McGuinness, D. et al. Socio-economic status is associated with epigenetic differences in the pSoBid cohort. Int. J. Epidemiol. 41, 151–160 (2012).

    PubMed  Google Scholar 

  159. Yousefzadeh, M. J. et al. Fisetin is a senotherapeutic that extends health and lifespan. EBioMedicine 36, 18–28 (2018).

    PubMed  PubMed Central  Google Scholar 

  160. Singh, S., Garg, G., Singh, A. K., Bissoyi, A. & Rizvi, S. I. Fisetin, a potential caloric restriction mimetic, attenuates senescence biomarkers in rat erythrocytes. Biochem. Cell Biol. 97, 480–487 (2019).

    CAS  PubMed  Google Scholar 

  161. Shi, Y. S. et al. Fisetin attenuates metabolic dysfunction in mice challenged with a high-fructose diet. J. Agric. Food Chem. 66, 8291–8298 (2018).

    CAS  PubMed  Google Scholar 

  162. Bondonno, N. P. et al. Fruit intake and abdominal aortic calcification in elderly women: a prospective cohort study. Nutrients 8, 159 (2016).

    PubMed  PubMed Central  Google Scholar 

  163. Xu, X. et al. Effects of dietary apple polyphenols supplementation on hepatic fat deposition and antioxidant capacity in finishing pigs. Animals 9, 937 (2019).

    PubMed Central  Google Scholar 

  164. Proshkina, E. et al. Geroprotective and radioprotective activity of quercetin, (-)-epicatechin, and ibuprofen in Drosophila melanogaster. Front. Pharmacol. 7, 505 (2016).

    PubMed  PubMed Central  Google Scholar 

  165. Yang, H., Song, Y., Liang, Y. N. & Li, R. Quercetin treatment improves renal function and protects the kidney in a rat model of adenine-induced chronic kidney disease. Med. Sci. Monit. 24, 4760–4766 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Stenvinkel, P. & Haase, V. H. Inflamed fat and mitochondrial dysfunction in end-stage renal disease links to hypoxia-could curcumin be of benefit? Nephrol. Dial. Transplant. 32, 909–912 (2017).

    CAS  PubMed  Google Scholar 

  167. Takano, K., Tatebe, J., Washizawa, N. & Morita, T. Curcumin inhibits age-related vascular changes in aged mice fed a high-fat diet. Nutrients 10, E1476 (2018).

    PubMed  Google Scholar 

  168. La Fata, G., Seifert, N., Weber, P. & Mohajeri, M. H. Vitamin E supplementation delays cellular senescence in vitro. Biomed. Res. Int. 2015, 563247 (2015).

    PubMed  PubMed Central  Google Scholar 

  169. Malavolta, M. et al. Changes in Zn homeostasis during long term culture of primary endothelial cells and effects of Zn on endothelial cell senescence. Exp. Gerontol. 99, 35–45 (2017).

    CAS  PubMed  Google Scholar 

  170. Jankowska, M., Rutkowski, B. & Dębska-Ślizień, A. Vitamins and microelement bioavailability in different stages of chronic kidney disease. Nutrients 9, E282 (2017).

    PubMed  Google Scholar 

  171. Galvan, D. L., Green, N. H. & Danesh, F. R. The hallmarks of mitochondrial dysfunction in chronic kidney disease. Kidney Int. 92, 1051–1057 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Li, Q., Zhang, A., Xing, C. & Yuan, Y. Disruption of mitochondrial homeostasis in chronic kidney disease: a mini-review. Histol. Histopathol. 34, 835–842 (2019).

    CAS  PubMed  Google Scholar 

  173. Sergi, D. et al. Mitochondrial (dys)function and insulin resistance: from pathophysiological molecular mechanisms to the impact of diet. Front. Physiol. 10, 532 (2019).

    PubMed  PubMed Central  Google Scholar 

  174. Serrano, J. C. E., Cassanye, A., Martín-Gari, M., Granado-Serrano, A. B. & Portero-Otín, M. Effect of dietary bioactive compounds on mitochondrial and metabolic flexibility. Diseases 4, 14 (2016).

    PubMed Central  Google Scholar 

  175. Mafra, D. et al. Bioactive food and exercise in chronic kidney disease: targeting the mitochondria. Eur. J. Clin. Invest. 48, e13020 (2018).

    PubMed  Google Scholar 

  176. Martinez Cantarin, M. et al. Uremia induces adipose tissue inflammation and muscle mitochondrial dysfunction. Nephrol. Dial. Transplant. 32, 943–951 (2017).

    PubMed  Google Scholar 

  177. Liu, C. et al. Reduced skeletal muscle expression of mitochondrial-derived peptides humanin and MOTS-C and Nrf2 in chronic kidney disease. Am. J. Physiol. Renal Physiol. 317, F1122–F1131 (2019).

    CAS  PubMed  Google Scholar 

  178. Clark, A. & Mach, N. The crosstalk between the gut microbiota and mitochondria during exercise. Front. Physiol. 8, 319 (2017).

    PubMed  PubMed Central  Google Scholar 

  179. Mafra, D., Borges, N. A., Lindholm, B. & Stenvinkel, P. Mitochondrial dysfunction and gut microbiota imbalance: an intriguing relationship in chronic kidney disease. Mitochondrion. 47, 206–209 (2019).

    CAS  PubMed  Google Scholar 

  180. Schrauwen, P., Schrauwen-Hinderling, V., Hoeks, J. & Hesselink, M. K. Mitochondrial dysfunction and lipotoxicity. Biochim. Biophys. Acta 1801, 266–271 (2010).

    CAS  PubMed  Google Scholar 

  181. Yuzefovych, L., Wilson, G. & Rachek, L. Different effects of oleate vs. palmitate on mitochondrial function, apoptosis, and insulin signaling in L6 skeletal muscle cells: role of oxidative stress. Am. J. Physiol. Endocrinol. Metab. 299, E1096–E1105 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  182. Lanza, I. R. et al. Influence of fish oil on skeletal muscle mitochondrial energetics and lipid metabolites during high-fat diet. Am. J. Physiol. Endocrinol. Metab. 304, E1391–E1403 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  183. Motawi, T. M. K., Hashem, R. M., Rashed, L. A. & El-Razek, S. M. A. Comparative study between the effect of the peroxisome proliferator activated receptor-α ligands fenofibrate and n-3 polyunsaturated fatty acids on activation of 5′-AMP-activated protein kinase-α1 in high-fat fed rats. J. Pharm. Pharmacol. 61, 1339–1346 (2009).

    CAS  PubMed  Google Scholar 

  184. Sun, X. & Zemel, M. B. Leucine modulation of mitochondrial mass and oxygen consumption in skeletal muscle cells and adipocytes. Nutr. Metab. 6, 26 (2009).

    Google Scholar 

  185. Sun, X. & Zemel, M. B. Leucine and calcium regulate fat metabolism and energy partitioning in murine adipocytes and muscle cells. Lipids 42, 297–305 (2017).

    Google Scholar 

  186. Sharma, S. & Black, S. M. Carnitine homeostasis, mitochondrial function, and cardiovascular disease. Drug Discov. Today Dis. Mech. 6, e31–e39 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Steiber, A., Kerner, J. & Hoppel, C. L. Carnitine: a nutritional, biosynthetic, and functional perspective. Mol. Asp. Med. 25, 455–473 (2004).

    CAS  Google Scholar 

  188. Yang, S.-K. et al. Effect of L-carnitine therapy on patients in maintenance hemodialysis: a systematic review and meta-analysis. J. Nephrol. 27, 317–329 (2014).

    CAS  PubMed  Google Scholar 

  189. Makrecka-Kuka, M. et al. Trimethylamine N-oxide impairs pyruvate and fatty acid oxidation in cardiac mitochondria. Toxicol. Lett. 267, 32–38 (2017).

    CAS  PubMed  Google Scholar 

  190. Vallance, H. D. et al. Marked elevation in plasma trimethylamine-N-oxide (TMAO) in patients with mitochondrial disorders treated with oral l-carnitine. Mol. Genet. Metab. Rep. 15, 130–133 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  191. Tang, W. H. et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N. Engl. J. Med. 368, 1575–1584 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  192. Jonsson, A. L. & Bäckhed, F. Role of gut microbiota in atherosclerosis. Nat. Rev. Cardiol. 14, 79–87 (2017).

    CAS  PubMed  Google Scholar 

  193. Wang, Z. et al. Impact of chronic dietary red meat, white meat, or non-meat protein on trimethylamine N-oxide metabolism and renal excretion in healthy men and women. Eur. Heart J. 40, 583–594 (2019).

    CAS  PubMed  Google Scholar 

  194. Zhong, V. W. et al. Associations of processed meat, unprocessed red meat, poultry, or fish intake with incident cardiovascular disease and all-cause mortality. JAMA Intern. Med. 80, 503–512 (2020).

    Google Scholar 

  195. Gross, J. L. et al. Effect of a chicken-based diet on renal function and lipid profile in patients with type 2 diabetes: a randomized crossover trial. Diabetes Care 25, 645–651 (2002).

    PubMed  Google Scholar 

  196. Bolati, D., Shimizu, H., Yisireyili, M., Nishijima, F. & Niwa, T. Indoxyl sulfate, a uremic toxin, downregulates renal expression of Nrf2 through activation of NF-κB. BMC Nephrol. 14, 56 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  197. Enoki, Y. et al. Potential therapeutic interventions for chronic kidney disease-associated sarcopenia via indoxyl sulfate-induced mitochondrial dysfunction. J. Cachexia Sarcopenia Muscle 8, 735–747 (2017).

    PubMed  PubMed Central  Google Scholar 

  198. Gao, Z. et al. Butyrate improves insulin sensitivity and increases energy expenditure in mice. Diabetes 58, 1509–1517 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  199. Bajpai, P., Darra, A. & Agrawal, A. Microbe-mitochondrion crosstalk and health: an emerging paradigm. Mitochondrion 39, 20–25 (2018).

    CAS  PubMed  Google Scholar 

  200. Tirosh, O., Levy, E. & Reifen, R. High selenium diet protects against TNBS-induced acute inflammation, mitochondrial dysfunction, and secondary necrosis in rat colon. Nutrition 23, 878–886 (2017).

    Google Scholar 

  201. Zhang, C. et al. Selenium triggers Nrf2-mediated protection against cadmium-induced chicken hepatocyte autophagy and apoptosis. Toxicol. In Vitro 44, 349–356 (2017).

    CAS  PubMed  Google Scholar 

  202. Lagouge, M. et al. Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1α. Cell 127, 1109–1122 (2006).

    CAS  PubMed  Google Scholar 

  203. Yuan, Y. et al. Activation of peroxisome proliferator activated receptor-g coactivator 1a ameliorates mitochondrial dysfunction and protects podocytes from aldosterone-induced injury. Kidney Int. 82, 771–789 (2012).

    CAS  PubMed  Google Scholar 

  204. Chang, Y. P. et al. Resveratrol inhibits NLRP3 inflammasome activation by preserving mitochondrial integrity and augmenting autophagy. J. Cell. Physiol. 230, 1567–1579 (2015).

    CAS  PubMed  Google Scholar 

  205. Hui, Y. et al. Resveratrol improves mitochondrial function in the remnant kidney from 5/6 nephrectomized rats. Acta Histochem. 119, 392–399 (2017).

    CAS  PubMed  Google Scholar 

  206. Den Hartogh, D. J. & Tsiani, E. Health benefits of resveratrol in kidney disease: evidence from in vitro and in vivo studies. Nutrients 11, 1624 (2019).

    CAS  Google Scholar 

  207. Wu, M. et al. Resveratrol delays polycystic kidney disease progression through attenuation of nuclear factor κB-induced inflammation. Nephrol. Dial. Transplant. 31, 1826–1834 (2016).

    CAS  PubMed  Google Scholar 

  208. Alvarenga, L. A. et al. Curcumin — a promising nutritional strategy for chronic kidney disease patients. J. Func. Foods 40, 715–721 (2018).

    CAS  Google Scholar 

  209. Correa, F. et al. Curcumin maintains cardiac and mitochondrial function in chronic kidney disease. Free. Radic. Biol. Med. 61, 119–12 (2013).

    CAS  PubMed  Google Scholar 

  210. Hernandez-Resendiz, S. et al. Cardioprotection by curcumin post-treatment in rats with established chronic kidney disease. Cardiovasc. Drugs Ther. 29, 111–120 (2015).

    CAS  PubMed  Google Scholar 

  211. Sudirman, S., Lai, C. S., Yan, Y. L., Yeh, H. I. & Kong, Z. L. Histological evidence of chitosan-encapsulated curcumin suppresses heart and kidney damages on streptozotocin-induced type-1 diabetes in mice model. Sci. Rep. 9, 15233 (2019).

    PubMed  PubMed Central  Google Scholar 

  212. Ghosh, S. S., Gehr, T. W. & Ghosh, S. Curcumin and chronic kidney disease (CKD): major mode of action through stimulating endogenous intestinal alkaline phosphatase. Molecules 19, 20139–20156 (2014).

    PubMed  PubMed Central  Google Scholar 

  213. Cuomo, F., Perugini, L., Marconi, E., Messia, M. C. & Lopez, F. Enhanced curcumin bioavailability through nonionic surfactant/caseinate mixed nanoemulsions. J. Food Sci. 84, 2584–2591 (2019).

    CAS  PubMed  Google Scholar 

  214. Vecchione, R. et al. Curcumin bioavailability from oil in water nano-emulsions: In vitro and in vivo study on the dimensional, compositional and interactional dependence. J. Control. Rel. 233, 88–100 (2016).

    CAS  Google Scholar 

  215. Chakraborty, M., Bhattacharjee, A. & Kamath, J. V. Cardioprotective effect of curcumin and piperine combination against cyclophosphamide-induced cardiotoxicity. Indian J. Pharmacol. 49, 65–70 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  216. Borlinghaus, J., Albrecht, F., Gruhlke, M. C., Nwachukwu, I. D. & Slusarenko, A. J. Allicin: chemistry and biological properties. Molecules 19, 12591–12618 (2014).

    PubMed  PubMed Central  Google Scholar 

  217. Salehi, B. et al. Allicin and health: a comprehensive review. Trends Food Sci. Technol. 86, 502–516 (2019).

    CAS  Google Scholar 

  218. Supakul, L. et al. Protective effects of garlic extract on cardiac function, heart rate variability, and cardiac mitochondria in obese insulin-resistant rats. Eur. J. Nutr. 53, 919–928 (2014).

    PubMed  Google Scholar 

  219. Zhang, M. et al. Allicin decreases lipopolysaccharide-induced oxidative stress and inflammation in human umbilical vein endothelial cells through suppression of mitochondrial dysfunction and activation of Nrf2. Cell. Physiol. Biochem. 41, 2255–2267 (2017).

    CAS  PubMed  Google Scholar 

  220. García Trejo, E. M. A. et al. The beneficial effects of allicin in chronic kidney disease are comparable to losartan. Int. J. Mol. Sci. 18, 1980 (2017).

    PubMed Central  Google Scholar 

  221. Granata, S. et al. Mitochondria: a new therapeutic target in chronic kidney disease. Nutr. Metab. 12, 49 (2015).

    Google Scholar 

  222. Perez-Cruz, I., Carcamo, J. M. & Golde, D. W. Vitamin C inhibits FAS-induced apoptosis in monocytes and U937 cells. Blood 102, 336–343 (2013).

    Google Scholar 

  223. Hao, J. et al. Role of vitamin C in cardioprotection of ischemia/reperfusion injury by activation of mitochondrial KATP channel. Chem. Pharm. Bull. 64, 548–557 (2016).

    CAS  Google Scholar 

  224. D’Costa, M. R. et al. Oxalosis associated with high-dose vitamin C ingestion in a peritoneal dialysis patient. Am. J. Kidney Dis. 74, 417–420 (2019).

    PubMed  PubMed Central  Google Scholar 

  225. Chang, H., Wang, Y., Yin, X., Liu, X. & Xuan, H. Ethanol extract of propolis and its constituent caffeic acid phenethyl ester inhibit breast cancer cells proliferation in inflammatory microenvironment by inhibiting TLR4 signal pathway and inducing apoptosis and autophagy. BMC Complement. Altern. Med. 17, 471 (2017).

    PubMed  PubMed Central  Google Scholar 

  226. Kubiliene, L. et al. Comparison of aqueous, polyethylene glycol-aqueous and ethanolic propolis extracts: antioxidant and mitochondria modulating properties. BMC Complement. Altern. Med. 18, 165 (2018).

    PubMed  PubMed Central  Google Scholar 

  227. Silveira, M. A. D. et al. Effects of Brazilian green propolis on proteinuria and renal function in patients with chronic kidney disease: a randomized, double-blind, placebo-controlled trial. BMC Nephrol. 20, 140 (2019).

    PubMed  PubMed Central  Google Scholar 

  228. Nadia, B. H. et al. Disruption of mitochondrial membrane potential by ferulenol and restoration by propolis extract: antiapoptotic role of propolis. Acta Biol. Hung. 60, 385–398 (2009).

    PubMed  Google Scholar 

  229. Ulusoy, H. B., Öztürk, İ. & Sönmez, M. F. Protective effect of propolis on methotrexate-induced kidney injury in the rat. Ren. Fail. 38, 744–750 (2016).

    CAS  PubMed  Google Scholar 

  230. Pedruzzi, L. M., Stockler-Pinto, M. B., Leite, M. Jr. & Mafra, D. Nrf2-keap1 system versus NF-κB: the good and the evil in chronic kidney disease? Biochimie 94, 2461–2466 (2012).

    CAS  PubMed  Google Scholar 

  231. Franceschi, C. et al. Inflammaging: a new immune-metabolic viewpoint for age-related diseases. Nat. Rev. Endocrinol. 14, 576–590 (2018).

    CAS  PubMed  Google Scholar 

  232. Sato, Y. & Yanagita, M. Immunology of the ageing kidney. Nat. Rev. Nephrol. 15, 625–640 (2019).

    PubMed  Google Scholar 

  233. Schmitz, M. L., Weber, A., Roxlau, T., Gaestel, M. & Kracht, M. Signal integration, crosstalk mechanisms and networks in the function of inflammatory cytokines. Biochim. Biophys. Acta 1813, 2165–2175 (2011).

    CAS  PubMed  Google Scholar 

  234. Armutcu, F. Organ crosstalk: the potent roles of inflammation and fibrotic changes in the course of organ interactions. Inflamm. Res. 68, 825–839 (2019).

    CAS  PubMed  Google Scholar 

  235. Zhang, J. et al. Ageing and the telomere connection: an intimate relationship with inflammation. Ageing Res. Rev. 25, 55–69 (2016).

    CAS  PubMed  Google Scholar 

  236. Bordoni, A. et al. Dairy products and inflammation: a review of the clinical evidence. Crit. Rev. Food Sci. Nutr. 57, 2497–2525 (2017).

    CAS  PubMed  Google Scholar 

  237. Bolori, P. et al. Adherence to a healthy plant diet may reduce inflammatory factors in obese and overweight women-a cross-sectional study. Diabetes Metab. Syndr. 13, 2795–2802 (2019).

    PubMed  Google Scholar 

  238. Galiè, S. et al. Impact of nutrition on telomere health: systematic review of observational cohort studies and randomized clinical trials. Adv. Nutr. 11, 576–601 (2019).

    PubMed Central  Google Scholar 

  239. Hussain, T. et al. Oxidative stress and inflammation: what polyphenols can do for us? Oxid. Med. Cell Longev. 2016, 7432797 (2016).

    PubMed  PubMed Central  Google Scholar 

  240. Marx, W. et al. The effect of polyphenol-rich interventions on cardiovascular risk factors in haemodialysis: a systematic review and meta-analysis. Nutrients 9, 1345 (2017).

    PubMed Central  Google Scholar 

  241. Bellezza, I. et al. Nrf2-Keap1 signaling in oxidative and reductive stress. Bioch. Biophys. Acta Mol. Cell Res. 1865, 721–733 (2018).

    CAS  Google Scholar 

  242. Battino, M. et al. Nrf2 as regulator of innate immunity: a molecular Swiss army knife! Biotech. Adv. 36, 358–370 (2018).

    CAS  Google Scholar 

  243. Ahmed, S. M. U., Luo, L., Namani, A., Wang, X. J. & Tang, X. Nrf2 signaling pathway: pivotal roles in inflammation. Biochim. Biophys. Acta Mol. Basis Dis. 1863, 585–597 (2017).

    CAS  PubMed  Google Scholar 

  244. Dinkova-Kostova, A. T., Fahey, J. W., Kostov, R. V. & Kensler, T. W. KEAP1 and Done? Targeting the NRF2 Pathway with Sulforaphane. Trends Food Sci. Technol. 69, 257–269 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  245. Martini, S. et al. Integrative biology identifies shared transcriptional networks in CKD. J. Am. Soc. Nephrol. 25, 2559–2572 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  246. Wu, K. C., McDonald, P. R., Liu, J. & Klaassen, C. D. Screening of natural compounds as activators of the keap1-Nrf2 pathway. Planta Med. 80, 97–104 (2014).

    CAS  PubMed  Google Scholar 

  247. Smith, R. E. The effects of dietary supplements that overactivate the Nrf2/ARE system. Curr. Med. Chem. 27, 2077–2094 (2020).

    CAS  PubMed  Google Scholar 

  248. Davinelli, S., Willcox, C. & Scapagnini, G. Extending healthy ageing: nutrient sensitive pathway and centenarian population. Immun. Ageing. 9, 9 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  249. Stenvinkel, P. et al. Novel treatment strategies for chronic kidney disease: insights from the animal kingdom. Nat. Rev. Nephrol. 14, 265–284 (2018).

    PubMed  Google Scholar 

  250. Paunkov, A., Chartoumpekis, D. V., Ziros, P. G. & Sykiotis, G. P. A bibliometric review of the Keap1/Nrf2 pathway and its related antioxidant compounds. Antioxidants. 8, E353 (2019).

    PubMed  Google Scholar 

  251. Saraiva, J. A. et al. Effects of low protein diet on nuclear factor erythroid 2-related factor 2 gene expression in nondialysis chronic kidney disease patients. J. Ren. Nutr. 30, 46–52 (2020).

    Google Scholar 

  252. McClelland, R. et al. Accelerated ageing and renal dysfunction links lower socioeconomic status and dietary phosphate intake. Aging 8, 1135–1149 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  253. Rebholz, C. M. et al. DASH (dietary approaches to stop hypertension) diet and risk of subsequent kidney disease. Am. J. Kidney Dis. 68, 853–861 (2016).

    PubMed  PubMed Central  Google Scholar 

  254. Bomback, A. S. et al. Sugar-sweetened soda consumption, hyperuricemia, and kidney disease. Kidney Int. 77, 609–616 (2010).

    CAS  PubMed  Google Scholar 

  255. Yang, M. et al. Apigenin prevents metabolic syndrome in high-fructose diet-fed mice by Keap1-Nrf2 pathway. Biomed. Pharmacother. 105, 1283–1290 (2018).

    CAS  PubMed  Google Scholar 

  256. Woo, M., Kim, M., Noh, J. S. & Song, Y. O. Kimchi methanol extracts attenuate hepatic steatosis induced by high cholesterol diet in low-density lipoprotein receptor knockout mice through inhibition of endoplasmic reticulum stress. J. Funct. Foods 32, 218–222 (2017).

    CAS  Google Scholar 

  257. Shin, J. H. et al. Nrf2-Heme oxygenase-1 attenuates high-glucose-induced epithelial-to-mesenchymal transition of renal tubule cells by inhibiting ROS-mediated PI3K/Akt/GSK-3β Signaling. J. Diabetes Res. 2019, 2510105 (2019).

    PubMed  PubMed Central  Google Scholar 

  258. Axelsson, A. S. et al. Sulforaphane reduces hepatic glucose production and improves glucose control in patients with type 2 diabetes. Sci. Transl. Med. 9, eaah4477 (2017).

    PubMed  Google Scholar 

  259. Gigliotti, J. C. et al. GSTM1 deletion exaggerates kidney injury in experimental mouse models and confers the protective effect of cruciferous vegetables in mice and humans. J. Am. Soc. Nephrol. 31, 102–116 (2020).

    CAS  PubMed  Google Scholar 

  260. Zhang, W., Li, Y., Ding, H., Du, Y. & Wang, L. Hydrogen peroxide prevents vascular calcification induced ROS production by regulating Nrf-2 pathway. Ren. Fail. 38, 1099–1106 (2016).

    CAS  PubMed  Google Scholar 

  261. Dai, L., Qureshi, A. R., Witasp, A., Lindholm, B. & Stenvinkel, P. Early vascular ageing and cellular senescence in chronic kidney disease. Comput. Struct. Biotechnol. J. 17, 721–729 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  262. Kunnumakkara, A. B. et al. Chronic diseases, inflammation, and spices: how are they linked? J. Transl. Med. 16, 14 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  263. Tsui, P.-F., Lin, C.-S., Ho, L.-J. & Lai, J.-H. Spices and atherosclerosis. Nutrients 10, 1724 (2018).

    PubMed Central  Google Scholar 

  264. Nilius, B. & Appendino, G. Spices: the savory and beneficial science of pungency. Rev. Physiol. Biochem. Pharmacol. 164, 1–76 (2013).

    CAS  PubMed  Google Scholar 

  265. Kocaadam, B. & Şanlier, N. Curcumin, an active component of turmeric (Curcuma Longa), and its effects on health. Crit. Rev. Food Sci. Nutr. 57, 2889–2895 (2017).

    CAS  PubMed  Google Scholar 

  266. White, C. M., Pasupuleti, V., Roman, Y. M., Li, Y. & Hernandez, A. V. Oral Turmeric/curcumin effects on inflammatory markers in chronic inflammatory diseases: a systematic review and meta-analysis of randomized controlled trials. Pharmacol. Res. 146, 104280 (2019).

    PubMed  Google Scholar 

  267. Ali, B. H. et al. Curcumin ameliorates kidney function and oxidative stress in experimental chronic kidney disease. Basic Clin. Pharmacol. Toxicol. 122, 65–73 (2018).

    CAS  PubMed  Google Scholar 

  268. Jiménez-Osorio, A. S. et al. The effect of dietary supplementation with curcumin on redox status and Nrf2 activation in patients with nondiabetic or diabetic proteinuric chronic kidney disease: a pilot study. J. Ren. Nutr. 26, 237–44 (2016).

    PubMed  Google Scholar 

  269. Ghosh, S. S., He, H., Wang, J., Gehr, T. W. & Ghosh, S. Curcumin-mediated regulation of intestinal barrier function: the mechanism underlying its beneficial effects. Tissue Barriers 6, e1425085 (2018).

    PubMed  PubMed Central  Google Scholar 

  270. Hami, M. et al. The effect of curcumin in prevention of contrast nephropathy following coronary angiography or angioplasty in CKD patients. Iran. J. Kidney Dis. 13, 304–309 (2019).

    PubMed  Google Scholar 

  271. Weir, M. A. et al. Micro-particle curcumin for the treatment of chronic kidney disease-1: study protocol for a multicenter clinical trial. Can. J. Kidney Health Dis. 5, (2018).

  272. Wang, Y. et al. Epigallocatechin-3-gallate attenuates oxidative stress and inflammation in obstructive nephropathy via NF-κB and Nrf2/HO-1 signalling pathway regulation. Basic Clin. Pharmacol. Toxicol. 117, 164–172 (2015).

    CAS  PubMed  Google Scholar 

  273. Jhee, H. J. et al. Effects of coffee intake on incident chronic kidney disease: a community-based prospective cohort study. Am. J. Med. 131, 1482–1490 (2018).

    CAS  PubMed  Google Scholar 

  274. Liang, N. & Kitts, D. D. Role of chlorogenic acids in controlling oxidative and inflammatory stress conditions. Nutrients 8, 16 (2015).

    PubMed Central  Google Scholar 

  275. Priftis, A., Angeli-Terzidou, A. E., Veskoukis, A. S., Spandidos, D. A. & Kouretas, D. Cell-specific and roasting-dependent regulation of the Keap1/Nrf2 pathway by coffee extracts. Mol. Med. Rep. 17, 8325–8331 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  276. Rassaf, T. et al. Vasculoprotective effects of dietary cocoa flavanols in patients on hemodialysis: a double-blind, randomized, placebo-controlled trial. Clin. J. Am. Soc. Nephrol. 11, 108–118 (2016).

    CAS  PubMed  Google Scholar 

  277. Hariri, M. & Ghiasvand, R. Cinnamon and chronic diseases. Adv. Exp. Med. Biol. 929, 1–24 (2016).

    CAS  PubMed  Google Scholar 

  278. Nabavi, S. F. et al. Nrf2 as molecular target for polyphenols: a novel therapeutic strategy in diabetic retinopathy. Crit. Rev. Clin. Lab. Sci. 53, 293–312 (2016).

    CAS  PubMed  Google Scholar 

  279. Kalt, W. et al. Recent research on the health benefits of Blueberries and Their Anthocyanins. Adv. Nutr. 11, 224–236 (2019).

    PubMed Central  Google Scholar 

  280. Cassidy, A. et al. High anthocyanin intake is associated with a reduced risk of myocardial infarction in young and middle-aged women. Circulation 127, 188–196 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  281. Moriwaki, S. et al. Delphinidin, one of the major anthocyanidins, prevents bone loss through the inhibition of excessive osteoclastogenesis in osteoporosis model mice. PLoS ONE 9, e97177 (2014).

    PubMed  PubMed Central  Google Scholar 

  282. Tang, J. S. et al. Bioavailable blueberry-derived phenolic acids at physiological concentrations enhance Nrf2-regulated antioxidant responses in human vascular endothelial cells. Mol. Nutr. Food Res. 62, 1–7 (2018).

    Google Scholar 

  283. Pan, J. et al. Pterostilbene, a bioactive component of blueberries, alleviates renal fibrosis in a severe mouse model of hyperuricemic nephropathy. Biomed. Pharmacother. 109, 1802–1808 (2018).

    PubMed  Google Scholar 

  284. Kelley, N., Jeltema, D., Duan, Y. & He, Y. The NLRP3 inflammasome: an overview of mechanisms of activation and regulation. Int. J. Mol. Sci. 20, 3328 (2019).

    CAS  PubMed Central  Google Scholar 

  285. Komada, T. & Muruve, D. A. The role of inflammasomes in kidney disease. Nat. Rev. Nephrol. 15, 501–520 (2019).

    PubMed  Google Scholar 

  286. Alvarenga, L. et al. Can nutritional interventions modulate the activation of the NLRP3 inflammasome in chronic kidney disease? Food Res. Int. https://doi.org/10.1053/j.jrn.2020.01.022 (2020).

  287. Hennig, P. et al. The crosstalk between Nrf2 and inflammasomes. Int. J. Mol. Sci. 19, 562 (2018).

    PubMed Central  Google Scholar 

  288. Lundberg, J. O., Weitzberg, E. & Gladwin, M. T. The nitrate-nitrite-nitric oxide pathway in physiology and therapeutics. Nat. Rev. Drug Discov. 7, 156–167 (2008).

    CAS  PubMed  Google Scholar 

  289. Senkus, K. E. & Crowe-White, K. M. Influence of mouth rinse use on the enterosalivary pathway and blood pressure regulation: a systematic review. Crit. Rev. Food Sci. Nutr. 2019, 1–13 (2019).

    Google Scholar 

  290. Bonilla, O. D. A. et al. Dietary nitrate from beetroot juice for hypertension: a systematic review. Biomolecules 8, E134 (2018).

    Google Scholar 

  291. Bryan, N. S. Functional nitric oxide nutrition to combat cardiovascular disease. Curr. Atheroscler. Rep. 20, 21 (2018).

    PubMed  Google Scholar 

  292. Sweazea, K. L., Johnston, C. S., Miller, B. & Gumpricht, E. Nitrate-rich fruit and vegetable supplement reduces blood pressure in normotensive healthy young males without significantly altering flow-mediated vasodilation: a randomized, double-blinded, controlled trial. J. Nutr. Metab. 2018, 1729653 (2018).

    PubMed  PubMed Central  Google Scholar 

  293. Edwards, M., Czank, C., Woodward, G. M., Cassidy, A. & Kay, C. D. Phenolic metabolites of anthocyanins modulate mechanisms of endothelial function. J. Agric. Food Chem. 63, 2423–2431 (2015).

    CAS  PubMed  Google Scholar 

  294. Curtis, P. P. et al. Blueberries improve biomarkers of cardiometabolic function in participants with metabolic syndrome-results from a 6-month, double-blind, randomized controlled trial. Am. J. Clin. Nutr. 109, 1535–1545 (2019).

    PubMed  PubMed Central  Google Scholar 

  295. Lundberg, J. O., Carlström, M. & Weitzberg, E. Metabolic effects of dietary nitrate in health and disease. Cell Metab. 28, 9–22 (2018).

    CAS  PubMed  Google Scholar 

  296. El Gamal, A. A. et al. Beetroot (Beta vulgaris L.) extract ameliorates gentamicin-induced nephrotoxicity associated oxidative stress, inflammation, and apoptosis in rodent model. Mediators Inflamm. 2014, 983952 (2014).

    PubMed  PubMed Central  Google Scholar 

  297. Bahadoran, Z. et al. Association between dietary intakes of nitrate and nitrite and the risk of hypertension and chronic kidney disease: Tehran Lipid and Glucose Study. Nutrients 21, E811 (2016).

    Google Scholar 

  298. Kemmner, S. et al. Dietary nitrate load lowers blood pressure and renal resistive index in patients with chronic kidney disease: a pilot study. Nitric Oxide 64, 7–15 (2017).

    CAS  PubMed  Google Scholar 

  299. Lobel, L., Cao, Y.G., Fenn, K., Glickman, J.N. & Garrett, W.S. Diet posttranslationally modifies the mouse gut microbial proteome to modulate renal function. Science 369, 1518–1524 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

D.M. receives support from Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq, grant no. 302034/2018-8) and Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ, grant no. E-26/202.24/2019). P.S. receives funding from the Strategic Research Program in Diabetes at Karolinska Institutet (Swedish Research Council grant no. 2009-1068), the European Union’s Horizon 2020 research and innovation Program under the Marie Skłodowska-Curie grant agreement no. 722609; International Network for Training on Risks of Vascular Intimal Calcification and roads to Regression of Cardiovascular Disease (INTRICARE). Baxter Novum is the result of a grant from Baxter Healthcare to the Karolinska Institutet.

Author information

Authors and Affiliations

Authors

Contributions

D.M., N.A.B., B.L., P.G.S. and P.S. researched the data for the article. All authors contributed to discussions of the content, wrote the text and reviewed or edited the manuscript before submission.

Corresponding author

Correspondence to Peter Stenvinkel.

Ethics declarations

Competing interests

B.L.’s research is funded by Baxter Healthcare. P.S. is on the scientific advisory boards of REATA, Baxter Healthcare and AstraZeneca. P.G.S. is funded through PhD studentships supported by 4D Pharma and Constant Pharma. The other authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Nephrology thanks N. Vaziri, A. Cupisti and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

INTRICARE: www.intricare.eu

Supplementary information

Glossary

Uraemic toxins

Biologically active compounds that are removed from the blood by healthy kidneys but can accumulate and exert toxic effects in the setting of kidney dysfunction.

Foodomics

The application of omics technologies to study food and nutrition.

Planetary health

The health of human civilization and the state of the natural systems on which it depends.

Methylome

The complement of methylation-based modifications in a genome or in a particular cell.

Diseaseome

The network of genes and pathways that is associated with human disease.

Osmoprotectant

Organic molecules, such as betaine, sugars and amino acids, that have neutral charge and maintain the integrity of cells exposed to osmotic stress.

One-carbon metabolism

A metabolic process that interlinks the methionine and folate cycles, which provide methyl groups for synthesis of DNA and maintenance of the epigenetic landscape.

Prebiotics

Non-digestible compounds in food that stimulate the growth of beneficial microorganisms such as bacteria and fungi.

Probiotics

Live microorganisms such as bacteria and yeasts that are thought to have health benefits when consumed.

Synbiotics

Dietary supplements that contain a combination of prebiotics and probiotics.

Salutogenic

Factors that maintain and promote human health.

Eryptosis

A type of programmed cell death that occurs in erythrocytes.

Nutraceuticals

Products derived from food that provide health benefits.

Senotherapeutics

Agents that target senescent cells, such as geroprotectors (which prevent or reverse the senescent state), senescence-associated secretory phenotype inhibitors, senolytics (which induce the death of senescent cells), senomorphics (which suppress senescent phenotypes without killing cells) and gene therapy strategies (which increase resistance to ageing).

Inflammageing

Inflammation that occurs during ageing or age-related diseases.

Blue zones

Regions in the world where people have high life expectancy, such as Okinawa (Japan), Loma Linda (USA), Sardinia (Italy), Nicoya (Costa Rica) and Icaria (Greece).

Kimchi

A traditional, heavily seasoned Korean dish made with vegetables such as napa cabbage and radish that are fermented by mainly lactic acid bacteria.

Natto

A traditional Japanese dish made with soybeans that are fermented by Bacillus subtilis.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mafra, D., Borges, N.A., Lindholm, B. et al. Food as medicine: targeting the uraemic phenotype in chronic kidney disease. Nat Rev Nephrol 17, 153–171 (2021). https://doi.org/10.1038/s41581-020-00345-8

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41581-020-00345-8

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing